ArticlePDF Available

Low-dose Light Therapy on Host Immune Response: Physiological Effects and Mechanisms of Action

Authors:

Abstract

The effect of low-dose light (LDL) therapy, commonly using red and near infrared (NIR) light (600-1 100 nm), has gained attention in recent years as a relatively noninvasive technique in modulating the tissue metabolic system, nervous system, blood circulation system and immune system. The progress in the basic science fields of bioenergetics and photobiology has propelled LDL into the therapeutic revolution. The immune cells including macrophages, mast cells, neutrophils and lymphocytes as responder cells by LDL have been studied in the animals and humans with producing cytokines and protective proteins. The paper will review the mechanisms of immune action of LDL at the molecular, cellular, and tissue levels on mammalian.
LowdoseLightTherapyonHostImmuneResponse:
PhysiologicalEffectsandMechanismsofAction
*
CHANGHao-Cai
1)
,ZHANGZhen-Zhen
2)
,LIULei
1)**
(
1) MOEKeyLaboratoryofLaserLifeScience&InstituteofLaserLifeScience,CollegeofBiophotonics,
SouthChinaNormalUniversity,Guangzhou
510631,China;
2) InstituteofHumanVirology,ZhongshanSchoolofMedicine,SunYat
-
senUniversityNorthCampus,Guangzhou
510080,China)
Abstract Theeffectoflow-doselight(LDL)therapy,commonlyusingredandnearinfrared(NIR)light(600-1100nm),hasgained
attentioninrecentyearsasarelativelynoninvasivetechniqueinmodulatingthetissuemetabolicsystem,nervoussystem,blood
circulationsystemandimmunesystem.TheprogressinthebasicsciencefieldsofbioenergeticsandphotobiologyhaspropelledLDL
intothetherapeuticrevolution.Theimmunecellsincludingmacrophages,mastcells,neutrophilsandlymphocytesasrespondercellsby
LDLhavebeenstudiedintheanimalsandhumanswithproducingcytokinesandprotectiveproteins.Thepaperwillreviewthe
mechanismsofimmuneactionofLDLatthemolecular,cellular,andtissuelevelsonmammalian.
Keywords Low-doselight,inflammatoryreaction,lymphedema,oropharyngealmucositis,radiationdermatitis,anti-infectionand
anti-tumor
DOI:10.16476/j.pibb.2017.0275
*ThisworkwassupportedbygrantsfromTheNationalNaturalScience
FoundationofChina(61361160414,31470072,61405061)andtheNatural
ScienceFoundationofGuangdongProvince,China(2014A030313419).
**Correspondingauthor.
Tel:86-20-85211436,E-mail:liulei@scnu.edu.cn
Received:July13,2017Accepted:October19,2017
化学物理进展
ProgressinBiochemistryandBiophysics
2017,44(12):1074~1082
www.pibb.ac.cn
Since1960s,Mester
[1]
discoveredthebiological
effectsoflow-doselight(LDL)actingonbiological
tissue,LDLhadgainedattentionbymoreandmore
researchersasanovelscientificapproach,which
inducednonthermalandnondestructivebiological
reactions,fortherapeuticapplicationsinavarietyof
experimentalconditions.Patients,researchersand
cliniciansaroundtheworldaredevotingattentionto
thepotentialtherapeuticapplicationsofLDLin
immunityandothermedicalfieldsthathave
traditionallyhadalimitedtherapeuticcontributionto
patientcare.
Recently,theuseofLDLhasextendedbeyond
therealmsofwoundhealingandpain,andrecent
researchsupportsitspotentialapplicationsin
neurodegenerativediseases
[2
-
3]
,type2diabetes
[4]
,
osteogenicdifferentiation
[5]
andthrombocytopenia
[6
-
7]
.
However,theexactmechanismsofthoseeffects
inducedbyLDLarepoorlyunderstood,butthe
mechanismisprobablytobephotochemicallyrelated.
Karu,apioneerintheLDLfield,proposedthat
cytochromecoxidase(CcO)wasthephotoacceptor
andsignaltransducer
[8
-
9]
,whichaffectedthe
mitochondrialelectrontransportsystem
[10]
andthe
biologicalregulationofreactiveoxygenspecies
(ROS)
[11
-
14]
,adenosinetriphosphate(ATP)
[15]
,nitric
oxide(NO)
[16
-
17]
andintracellularCa
2+[18
-
19]
,andfurther
affectedtheailmentprocessincludinginflammation
andcytokineandgrowthfactorrelease(Figure1).The
articlesummarizestheavailableliteratureon
molecularmechanismsoftheprotectiveorenhancing
好才,等:功率法对疫应影响
2017;44(12)
1ReducedinflammatoryreactionbyLDL
1.1 LDLregulatesthesecretionofcytokines
Inflammatoryreactionisthephysiological
reactioncausedbythestimulationoftrauma,bleeding
orpathogeninfection.Itistheinnateimmune
defensivereactionofimmunecellsandinflammatory
factors.Overtheyears,severalstudiesonhumansand
animalshaveshownthatLDLhasmodulatoryeffects
oninflammatorymarkersofIL-1,IL-6,IL-8,TNF-
andprostaglandinE2(PGE2),andrelievesthe
inflammatoryprocess(edema,necrosis,neutrophilcell
influx,hemorrhagicformation).AccordingtoChang
etal.
[20]
,inflammatorysymptomsarecausedbypro-
inflammatorycytokines,suchasIL-1,IL-6and
TNF-.StudybydeAlmeidaandcolleagues
[21]
reportedLDLsignificantlydecreasedtheinflammatory
mediatorlevelsofIL-1,IL-6andTNF-inacute
skeletalmuscleinjury.Similarly,LDLcouldreduced
thosecytokineproductioninthepathophysiologyof
osteoarthritis(OA)
[22]
.25J/cm
2
LDLalsodecreasedthe
levelofpro-inflammatorycytokinesofTNF-,IL-1,
andIL-8inrheumatoidarthritissynoviocytes
[23]
.
Additionally,thetrauma-inducedpro-inflammatory
stateassessedbyIL-6andIL-10waspreventedLDL
[24]
.
Inparallel,LDLpreventedtrauma-inducedreduction
inBDNFandVEGF,vascularremodelingand
fiber-proliferatingmarkers.Morerecently,LDLhas
beenshownveryinterestingeffectsonmodulation
ofcyclooxygenase2(COX-2).(880 10)nmLDL
decreasedtheinflammatorycellinfluxandmRNA
levelsofCOX-2justininitialphaseof Achilles
tendinitis
[25]
.COX-2mRNAexpressionswerealso
significantlydecreasedbytreatmentwith904nm
LDL
[26]
.Almeida etal
[27]
.alsofoundthat904nmLDL
in1.0Jgroupsignificantlydecreasesskeletalmuscle
effectsofLDLinanumberofpathogenicconditions
includinginflammatoryreaction,cancertherapy-
inducedcomplications(lymphedema,mucositisand
dermatitis),andanti-infectionandanti-tumoreffects.
Fig.1 ThemechanismmodelofLDL
Schematicdiagramshowstheredornearinfrared(NIR)lightisabsorbedbythephotoacceptors(e.g.cytochromecoxidase)localizedinmitochondria.
Duringtheprocess,ROSandATPproductionareincreased,NOisreleased,andintracellularCa
2+
concentration([Ca
2+
]
i
)iselevated.Theseresponses
mayultimatelyleadtochangesincellmorphologyandfunction
via
activatingsometranscriptionfactors[e.g.,nuclearfactor-
B(NF-
B),hypoxia
induciblefactor-1(HIF-1),activatorprotein-1(AP-1)andcAMP-responseelementbindingprotein(CREB)].
ROS
ATP
NO[Ca
2+
]
i
Messenger
Red/NIRlight
Nucleus
Cellmembrane
CREB
AP-1
HIF-1
NF-
B
Genetranscription
Cytokineandgrowthfactorproduction
Extracellularmatrixdeposition
Cellproliferation,migrationandadhesion
1075· ·
化学物理进展 Prog.Biochem.Biophys. 2017;44(12)
damagethroughlessCOX-2-derivedgeneexpression.
However,theprecisemechanismbywhichlight
affectsthecytokinesisnotyetknown.LDLprobably
modulatedthepro-inflammatorycytokinesbyreducing
theIL-1andCOX-2mRNAexpressionand
consequentlyreducedPGE2levelsbyreducingcell
migrationandthequantityofmacrophages,
neutrophils,andmastcellsintheinjuredtissue
[28
-
29]
.
Macrophagesandmastcellssecretethecytokineof
IL-1,whichinturnrecruitsCOX-2,anenzymewhich
convertsarachidonicacidintoPGE2
[30
-
31]
.Fromthe
aboveweseethatreducedinflammatoryreactionby
LDLmaydependnotonlyonthelightirradiation
parameters(wavelength,radiationdose),butalsoon
pathologicalconditionofthestudymodel.
1.2 LDLincreasesMMPsandPAactivity
Matrixmetalloproteinases(MMPs),whichare
consideredtodegradethecomponentsofthecomplex
extracellularmatrix,andplasminogenactivator(PA),
whichisimplicatedintheplasminogen-plasmin
proteolyticsystem,playakeyroleinextracellular
matrixdegradation,synthesisofkininandfibrinolysis
intheprocessofinflammation.Both660nmand780nm
verifiedbyCuryandcolleaguescoulddecreaseMMP2
activityinamodelofischemicskinflapinrats
[32]
.
AndMMP9activitywasdecreasedoninducedarthritis
inthetemporomandibularjointwith830nmLDL
treatment
[33]
.Additionally,severalhumanandanimal
studieshaveshownthatLDLwithredtoinfrared
wavelengthsreducesthereleaseofPA
[34
-
35]
.Thus,LDL
probablymodulatesPAactivitytodegradecell
adhesivemoleculesandextracellularmatrixproteins
[36]
throughactivationofMMPs
[37]
.Furthermore,
plasminogenactivatesthekinincascade via converting
prekallikreinintokallikrein
[38]
.
1.3 LDLmodulatestheimmunecellactivity
LDLalsomodulatestheactivityofmastcells,
macrophages,neutrophilsandlymphocytestoreduce
theinflammatoryprocess.RedLDLhasbeenshownto
inducethemast-celldegranulation
[39
-
40]
,leadingtothe
releaseofamultiplechemicalmediators(VEGF121,
VEGF165,VEGF189andVEGF206)
[41]
,whichare
relatedtovasodilationandvascularproliferation,and
canoptimizetheinflammatoryprocess
[42]
.Song etal.
[43]
reportedthatinratsLDLalteredthemacrophage
polarizationfromM1statetoM2state,whichdampens
theinflammatoryandadaptiveTh1responses
[44]
.Other
studiesobservedthatLDLreducedintheabsolute
numberofmacrophagesandneutrophilscompared
withtheinjurygroup
[22,45]
,resultingindecreaseof
secretionofpro-inflammatorycytokinesandenzymes
suchasIL-6andTNF-involvedindrivingthe
inflammatoryresponse
[46]
.Forlymphocytes,LDLcould
activatedirectlyitsproliferation invivo
[47
-
49]
,leadingto
secreteanti-inflammatorycytokineofIL-10,which
inhibitedtheproductionofpro-inflammatorycytokines
andpreventedmacrophageandneutrophilinfiltration
intotheinjury
[50]
.Additionally,thepresenceof
hemoglobinamplifiedtheproliferationeffectofLDL
irradiationonlymphocyteculture
[49]
.Hemoglobin
couldcatalyzefreeradicalformationinthepresenceof
hydrogenperoxideasintheFentonreaction
[51]
.LDLat
agivenwavelengthmaypromoteROSformationina
hemoglobinrichenvironment,andthenthegeneration
ofanoxidativeenvironmenthasastronginfluenceon
Tlymphocytes
[52]
.
2Reduced cancer therapyinduced
complicationsbyLDL
Notonlydrugresistancecausedbychemotherapy
andmoleculartargetedtherapyisamajorobstacleto
thecurrenttumortreatment
[53
-
55]
,butalsocancer
therapy-inducedcomplicationsareacommonclinical
problem.Inhumanresearches,LDLiswidelystudied
toamelioratecancertherapy-inducedcomplications.
Upperlimblymphedema,whichistheresultofthe
regionalaccumulationofamountsofprotein-rich
interstitialfluidcausedbyimpairedlymphdrainage
[56]
,
isacommoncomplicationofbreastcancersurgery.To
date,researchershavereportedthatLDLisbenefitfor
postmastectomylymphoedema
[57
-
59]
throughpresumably
increasingmicrocirculation
[60
-
61]
toreducetheexcessive
amountsoftissueproteinandfluid,andfinally
improvethelimbperformance.Inparticular,astudy
indicatedthatLDLwasoftenwithinhoursof
irradiationasanefficacytreatmentoflymphedema
[62]
.
However,themolecularmechanismsofLDLin
lymphoedematissueremainelusive.Atthe
microcirculatorylevel,thestimulatory/protective
effectsofLDLisachievedbymodulatingthe
angiogenicfactorproductionbylymphocytes
[63]
and
endothelialcells
[64]
in situ,thentoaccelerate
spontaneousangiogenesis
[65]
.
Oropharyngealmucositis(OM),knownasmost
painfulorallesions
[66]
,isamajorcomplicationofhead-
and-neckoncologictherapy
[67]
.LDLwasconfirmedto
beeffectiveincontrollingofOMcausedbyvarious
cancertherapies
[68
-
71]
.Studieshaveunambiguously
1076· ·
好才,等:功率法对疫应影响
2017;44(12)
demonstratedthatthemucositispathogenesisare
complexandassociatewithpro-inflammatory
cytokines
[72
-
73]
,microvascularinjury
[74]
,andextracellular
matrixalterations
[75
-
76]
.Silva etal.
[77]
showedthatLDL
increasedthelevelsofIL-10inbloodplasmaand
MMP-2insalivaon7thchemoradiotherapy-induced
OM.StudybyOton-Leite
[78]
demonstratedthatLDL
significantlyreducedsalivaryconcentrationofIL-6,
EGFandVEGFduringradiotherapysession.It
seeminglysuggestedthemechanismofLDL-reduced
theseverityofOMcausedbycancertherapywas
linkedtothemodulationofpro-oranti-inflammatory
cytokines,MMPsorgrowthfactors.Inananimal
modelofOM,studieshavereportedthatLDL
decreasedtheexpressionofCOX-2
[79]
,whichelicitsthe
synthesisofpro-inflammatoryprostaglandinsin
malignantandinflamedtissues,andreducesthe
infiltrationofneutrophilsininflamedtissues
[80]
,thus
furthersupportingtheanti-inflammatoryeffect.
Radiationdermatitis(RD)occursinamajorityof
breastcancerpatientswhoreceiveradiotherapyand
mayexhibitsymptomssuchasredness,itching,
dryness,andpeelingskin
[81]
.DeLand etal.
[82]
showed
thatLDLreducedtheincidenceofskinreactionsin
breastcancerpatientstreatedbyradiotherapy
postlumpectomy.Schindlandco-workers
[83
-
84]
demonstratedthatLDLhealedalong-lasting
radiotherapy-inducedskinulcer.Regardingthe
mechanismofaction,LDLhavebeendemonstratedto
induceneoangiogenesis via theactivationofERK/Sp1
pathway invitro
[64]
and invivo
[83]
,toaccelerate
collateralcirculationandenhancemicrocirculation
[85]
,
thentopossiblyimproveskincirculation
[86]
,andfinally
toreducetissuedamagecausedbyischemia
[87]
.An
alternativeexplanationofLDL-inducedneoangiogenesis
is via ROS
[88]
,whichleadtoincreasethelevelof
HIF-1
[89]
,thenregulatethetranscriptionofVEGF
[90
-
91]
.
Additionally,LDLcouldmodulatecertaincellular
proliferationandmigration
[92
-
94]
,andinducethe
secretionoffibroblastgrowthfactorfamilyinvolvedin
tissuerepair
[95]
.Altogether,thesefindingssuggesteda
beneficialeffectofLDLoncancertherapy-induced
complicationsandpatientsqualityoflifeincancer
patients.
3 Enhancedantiinfectionandantitumor
effectbyLDL
Recently,theeffectofLDL-inducedanti-
infectionwasfurtherconfirmedbyLu etal.
[96]
.They
showedthatLDLenhancedanti-infectionability in
vivo toimprovethemacrophagephagocyticactivity
throughRac1-mediatedsignalingpathway.
Simultaneously,Karunarathne etal.
[97]
showedthat
488-,515-,or595-nmwavelengthlightcouldinitiate
macrophagemigration.Theproductionof
pro-inflammatorycytokines(TNF-andIL-1)by
murineperitonealmacrophages invitro and invivo
wasraisedbyLDLaccompaniedwithincreasingthe
abilityofbacterialkilling
[98]
.Inneutrophils,LDLalso
enhancedtheabilitytokill Candidaalbicansvia the
generationofROS
[99]
.Inawoundinfectionmodel,it
wasdemonstratedLDLsignificantlydecreasedthe
incidencesofmicrobialflora(
Staphylococcusaureus
and Bacillussubtilis)comparedwithplaceboburns
[100]
,
andincreasedtheamountofbloodvessels,remodeled
thecollagenmatrix,andmaturedcollagenfibersin
infectedwounds
[101]
.
Theobviousparabolafeaturesofthebiological
effectofLDLoncellshavebeendemonstratedby
severalstudies
[102
-
103]
.Withanincreaseoflightoutput
energy,itsmoderatingactiononcellscanbeincreased
gradually,butwhenthelightoutputenergyexceedsa
certainthresholdvalue,theinhibitioneffectofLDL
emerges
[104]
.AccordingtoLuandcolleagues
[105]
,high
fluence,low-doselight(HF-LDL)wasreportedtokill
tumorcell,leadingtoactivatemacrophagestocreate
animmunememoryresponse.Afewmolecular
mechanismsrevealedthatHF-LDL-inducedapoptotic
tumorcellsenhancedthepro-inflammatorycytokines
(TNF-andNO)productioninmacrophage,through
upregulatingNF-Bactivity
[106
-
107]
.Thosestudiesmay
provideaneffectivetherapeuticapproachtoinducean
antitumorimmuneresponseafterHF-LDLtreatment.
4 Conclusion
Inconclusion,LDLhasstrongevidencesfor
manybeneficialeffectsoninflammatoryreaction,
cancertherapy-inducedcomplications,and
anti-infectionandanti-tumorinanimalmodelsand
humanpatients.Inthisreview,LDL-inducedthose
effectsmainlyinvolve4growthfactors(FGF,EGF,
TGF-andVEGF),5interleukins(IL-1,IL-4,IL-6,
IL-8andIL-10),5inflammatorycytokines(PGE2,
COX2,TNF-,MMPsandPA)and4immunecells
(macrophages,mastcells,neutrophilsand
lymphocytes).Themediatormoleculesinduced/
upregulatedbyLDLaresummarized(Table1).
However,theunderlyingmechanismsofthoseeffects
1077· ·
化学物理进展 Prog.Biochem.Biophys. 2017;44(12)
References
[1]MesterE,MesterAF,MesterA.Thebiomedicaleffectsoflaser
applicationonbiologicalsystems.LaserRev,1968,
1
:3
[2]GuX,LiuL,ShenQ,
etal
.PhotoactivationofERK/CREB/VMAT2
pathwayattenuatesMPP
+
-inducedneuronalinjuryinacellular
modelofParkinson'sdisease.CellularSignalling,2017,
37
:103
-
114
[3]MengCB,HeZY,XingD.Low-levellasertherapyrescues
dendriteatrophy
via
upregulatingBDNFexpression:implications
forAlzheimer'sdisease.JournalofNeuroscience,2013,
33
(33):
13505
-
13517
[4]JiangX,HuangL,XingD.PhotoactivationofDok1/ERK/
PPARgammasignalingaxisinhibitsexcessivelipolysisin
insulin-resistantadipocytes.CellularSignalling,2015,
27
(7):1265
-
1275
[5]FengJ,SunQ,LiuL,
etal
.PhotoactivationofTAZ
via
Akt/GSK3betasignalingpathwaypromotesosteogenic
differentiation.TheinternationalJournalofBiochemistry&Cell
Biology,2015,
66
:59
-
68
[6]ZhangQ,DongTT,LiPY,
etal
.Noninvasivelow-levellaser
therapyforthrombocytopenia.SciTranslMed,2016,
8
(349):
349ra101
[7]YangJK,ZhangQ,LiPY,
etal
.Low-levellighttreatment
amelioratesimmunethrombocytopenia.ScientificReports,2016,
6
:srep38238
[8]KaruTI.Mitochondrialsignalinginmammaliancellsactivatedby
redandnear-IRradiation.PhotochemistryandPhotobiology,2008,
84
(5):1091
-
1099
[9]KaruTI,PyatibratLV,MoskvinSV,
etal
.Elementaryprocesses
incellsafterlightabsorptiondonotdependonthedegreeof
polarization:Implicationsforthemechanismsoflaser
phototherapy.PhotomedicineandLaserSurgery,2008,
26
(2):
77
-
82
[10]YuW,NaimJO,McgowanM,
etal
.Photomodulationofoxidative
metabolismandelectronchainenzymesinratlivermitochondria.
PhotochemistryandPhotobiology,1997,
66
(6):866
-
871
[11]ZhangJT,XingD,GaoXJ.Low-powerlaserirradiationactivates
Srctyrosinekinasethroughreactiveoxygenspecies-mediated
signalingpathway.JournalofCellularPhysiology,2008,
217
(2):
518
-
528
[12]SunXG,WuSN,XingD.Thereactiveoxygenspecies-Src-Stat3
pathwayprovokesnegativefeedbackinhibitionofapoptosis
inducedbyhigh-fluencelow-powerlaserirradiation.FebsJournal,
2010,
277
(22):4789
-
4802
[13]ChenPJ,LuoXY,NiePP,
etal
.CQsynergisticallysensitizes
humancolorectalcancercellstoSN-38/CPT-11throughlysosomal
andmitochondrialapoptoticpathway
via
p53-ROScross-talk.Free
RadicalBioMed,2017,
104
:280
-
297
Mediatorclassification Molecules Actions/effects
Growthfactors NGF,GDNF,BDNF,FGF-1/2,bFGF,IGF-1/2,KGF,
PlGF,HGF,PDGF,TGF-
,TGF-
1/2,VEGF
Proliferation/Differentiation/Migration/Chemotaxis/
Angiogenesis/Anti-inflammatory/Extracellularmatrixsynthesis
Anti-inflammatorycytokinesIL-2,IL-4,IL-8,IL-10 Proliferation/Differentiation/Chemotaxis/Angiogenesis/
Immunologicalactivation
Pro-inflammatorycytokinesIL-1
,IL-1
,IL-6,IFN-
,TNF-
,PGE2,COX1,
COX2
Proliferation/Angiogenesis/Migration/Accelerate
inflammation/Immunologicalactivation
Chemokines
MCP-1(CCL2),MIP-1(CCL3),MIP-1(CCL4),CCR5
Chemotaxis/Immunologicalactivation
Celladhesionmolecules ICAM-1,VCAM-1,CD44,PECAM1,CTGF Proliferation/Differentiation/Angiogenesis/Migration/
Integrinactivation
Cellmatrixproteins MMP1,MMP2,MMP3,MMP9,MMP13,Types
and
collagen
Differentiation/Angiogenesis/Regeneration/
Tissueremodeling
Smallmolecules cGMP,cAMP,ATP,GSH,MDA,ROS,Ca
2+
,NO,H
+
Proliferation/Migration/Regulatingcellactivity/
Vasodilatation/Vasoconstriction
Others PA,PTH,Leukotriene,iNOS,LP,ET-1 Proliferation/Degradationoffibrin/Regulatingcellactivity/
Vasodilatation/Vasoconstriction
causedbyLDLarenotcompletelyunderstood.The
precisemolecularmechanismsarestillneededto
furtherexperimentsforpropellingLDLintothe
therapeuticrevolution.
Table1 MediatormoleculesassociatedwithLDL
Themediatormoleculesareinvolvedindifferentclassificationasnoted.Theactionsand/oreffectsofthosethemediatorsweresummarizedas
follows,whereastheyarenotcomprehensive.Rather,itprovidesinsightintothoseactivitiesassociatedwithLDL.Elementsofthetablewere
developedfromT.Sonis
[108]
.
1078· ·
好才,等:功率法对疫应影响
2017;44(12)
[14]AranyPR,ChoA,HuntTD,
etal
.Photoactivationofendogenous
latenttransforminggrowthfactor-beta1directsdentalstemcell
differentiationforregeneration.SciTranslMed,2014,
6
(238):
238ra69
[15]BlattA,Elbaz-GreenerGA,TubyH,
etal
.Low-levellasertherapy
tothebonemarrowreducesscarringandimprovesheartfunction
post-acutemyocardialinfarctioninthepig.Photomedicineand
LaserSurgery,2016,
34
(11):516
-
524
[16]GuerraFD,VieiraCP,OliveiraLP,
etal
.Low-levellasertherapy
modulatespro-inflammatorycytokinesafterpartialtenotomy.
LasersinMedicalScience,2016,
31
(4):759
-
766
[17]FarivarS,MalekshahabiT,ShiariR.Biologicaleffectsoflowlevel
lasertherapy.JournaloflasersinMedicalSciences,2014,
5
(2):
58
-
62
[18]MangTS,ManeshiMM,ShucardDW,
etal
.Effectsoflow-level
laserexposureoncalciumchannelsandintracellularreleasein
culturedastrocytes.LasersinSurgery&Medicine,2015,
47
(4):
383
[19]DeFreitasLF,HamblinMR.Proposedmechanismsof
photobiomodulationorlow-levellighttherapy.IeeeJSelTop
Quant,2016,
22
(3):pii7000417
[20]ChangXY,HeH,ZhuLP,
etal
.Protectiveeffectofapigeninon
Freund'scompleteadjuvant-inducedarthritisinrats
via
inhibiting
P2X7/NF-kappaBpathway. Chemico-BiologicalInteractions,
2015,
236
:41
-
46
[21]DeAlmeidaP,TomazoniSS,FrigoL,
etal
.Whatisthebest
treatmenttodecreasepro-inflammatorycytokinereleaseinacute
skeletalmuscleinjuryinducedbytraumainrats:low-levellaser
therapy,diclofenac,orcryotherapy?LasersinMedicalScience,
2014,
29
(2):653
-
658
[22]AlvesACA,VieiraRD,LealECP,
etal
.Effectoflow-levellaser
therapyontheexpressionofinflammatorymediatorsandon
neutrophilsandmacrophagesinacutejointinflammation.Arthritis
ResTher,2013,
15
(5):R116
[23]YamauraM,YaoM,YaroslavskyI,
etal
.Lowlevellighteffectson
inflammatorycytokineproductionbyrheumatoidarthritis
synoviocytes.LasersinSurgeryandMedicine,2009,
41
(4):282
-
290
[24]SilveiraPCL,SchefferDD,GlaserV,
etal
.Low-levellaser
therapyattenuatestheacuteinflammatoryresponseinducedby
muscletraumaticinjury.FreeRadicalRes,2016,
50
(5):503
-
513
[25]XavierM,DavidDR,DeSouzaRA,
etal
.Anti-inflammatory
effectsoflow-levellightemittingdiodetherapyonachilles
tendinitisinrats.LasersinSurgeryandMedicine,2010,
42
(6):
553
-
558
[26]LealECP,DeAlmeidaP,TomazoniSS,
etal
.Superpulsed
low-levellasertherapyprotectsskeletalmuscleofmdxmice
againstdamage,inflammationandmorphologicalchangesdelaying
dystrophyprogression.PloSOne,2014,
9
(3):e89453
[27]DeAlmeidaP,Lopes-MartinsRaB,TomazoniSS,
etal
.Low-
levellasertherapyimprovesskeletalmuscleperformance,decreases
skeletalmuscledamageandmodulatesmRNAexpressionof
COX-1andCOX-2inadose-dependentmanner.Photochemistry
andPhotobiology,2011,
87
(5):1159
-
1163
[28]DouradoDM,FaveroS,MatiasR,
etal
.Low-levellasertherapy
promotesvascularendothelialgrowthfactorreceptor-1expression
inendothelialandnonendothelialcellsofmicegastrocnemius
exposedtosnakevenom.PhotochemistryandPhotobiology,2011,
87
(2):418
-
426
[29]SilveiraLB,PratesRA,NovelliMD,
etal
.Investigationofmast
cellsinhumangingivafollowinglow-intensitylaserirradiation.
PhotomedicineandLaserSurgery,2008,
26
(4):315
-
321
[30]HardyMM,SeibertK,ManningPT,
etal
.Cyclooxygenase
2-dependentprostaglandinE-2modulatescartilageproteoglycan
degradationinhumanosteoarthritisexplants. Arthritisand
Rheumatism,2002,
46
(7):1789
-
1803
[31]MarsolaisD,CoteCH,FrenetteK.Neutrophilsandmacrophages
accumulatesequentiallyfollowingAchillestendoninjury.
JOrthopaedRes,2001,
19
(6):1203
-
1209
[32]CuryV,MorettiAIS,AssisL,
etal
.Lowlevellasertherapy
increasesangiogenesisinamodelofischemicskinflapinrats
mediatedbyVEGF,HIF-1alphaandMMP-2.JPhotochPhotobio
B,2013,
125
:164
-
170
[33]LemosGA,RissiR,PiresILD,
etal
.Low-levellasertherapy
stimulatestissuerepairandreducestheextracellularmatrix
degradationinratswithinducedarthritisinthetemporomandibular
joint.LasersinMedicalScience,2016,
31
(6):1051
-
1059
[34]VaninAA,DeMarchiT,TomazoniSS,
etal
.Pre-exerciseinfrared
low-levellasertherapy(810nm)inskeletalmuscleperformance
andpostexerciserecoveryinhumans,whatistheoptimaldose?A
randomized,double-blind,placebo-controlledclinicaltrial.
PhotomedicineandLaserSurgery,2016,
34
(10):473
-
482
[35]TakemaT,YamaguchiM,AbikoY.Reductionofplasminogen
activatoractivitystimulatedbylipopolysaccharidefromperiodontal
pathogeninhumangingivalfibroblastsbylow-energylaser
irradiation.LasersinMedicalScience,2000,
15
(1):35
-
42
[36]CastellinoFJ.Biochemistryofhumanplasminogen.Seminarsin
ThrombosisandHemostasis,1984,
10
(1):18
-
23
[37]WerbZ,MainardiCL,VaterCA,
etal
.Endogenousactivationof
latentcollagenasebyrheumatoidsynovialcells.Evidenceforarole
ofplasminogenactivator.TheNewEnglandJournalofMedicine,
1977,
296
(18):1017
-
1023
[38]VogtW.Kininformationbyplasmin,anindirectprocessmediated
byactivationofkallikrein.TheJournalofPhysiology,1964,
170
:153
-
166
[39]WangL,ZhangD,SchwarzW.TRPVchannelsinmastcellsasa
targetforlow-level-lasertherapy.Cells,2014,
3
(3):662
-
673
[40]YangWZ,ChenJY,YuJT,
etal
.Effectsoflowpowerlaser
irradiationonintracellularcalciumandhistaminereleaseinRBL-
2H3mastcells.PhotochemistryandPhotobiology,2007,
83
(4):
979
-
984
[41]PereiraMCMC,DePinhoCB,MedradoARP,
etal
.Influenceof
670nmlow-levellasertherapyonmastcellsandvascularresponse
ofcutaneousinjuries.JPhotochPhotobioB,2010,
98
(3):188
-
192
1079· ·
化学物理进展 Prog.Biochem.Biophys. 2017;44(12)
[42]ParaguassuGM,DeCastroICV,VasconcelosaRM,
etal
.Effect
ofLEDphototherapy(lambda630+/-20nm)onmastcells
duringwoundhealinginhypothyroid. ProcSPIE,2014,
8932
:893216-893216-8
[43]SongJW,LiK,LiangZW,
etal
.Low-levellaserfacilitates
alternativelyactivatedmacrophage/microgliapolarizationand
promotesfunctionalrecoveryaftercrushspinalcordinj\uryinrats.
ScientificReports,2017,
7
(1):620
[44]BouhlelMA,DerudasB,RigamontiE,
etal
.PPARgamma
activationprimeshumanmonocytesintoalternativeM2
macrophageswithanti-inflammatoryproperties.CellMetabolism,
2007,
6
(2):137
-
143
[45]DosSantosSA,AlvesAC,Leal-JuniorEC,
etal
.Comparative
analysisoftwolow-levellaserdosesontheexpressionof
inflammatorymediatorsandonneutrophilsandmacrophagesin
acutejointinflammation.LasersinMedicalScience,2014,
29
(3):
1051
-
1058
[46]KennedyA,FearonU,VealeDJ,
etal
.Macrophagesinsynovial
inflammation.FrontiersinImmunology,2011,
2
:52
[47]AvciP,GuptaA,SadasivamM,
etal
.Low-levellaser(light)
therapy(LLLT)inskin:stimulating,healing,restoring.Semin
CutanMedSurg,2013,
32
(1):41
-
52
[48]ManteifelV,BakeevaL,KaruT.Ultrastructuralchangesin
chondriomeofhumanlymphocytesafterirradiationwithHe-Ne
laser:Appearanceofgiantmitochondria.JPhotochPhotobioB,
1997,
38
(1):25
-
30
[49]StadlerI,EvansR,KolbB,
etal
.
Invitro
effectsoflow-levellaser
irradiationat660nmonperipheralbloodlymphocytes.Lasersin
SurgeryandMedicine,2000,
27
(3):255
-
261
[50]EfronPA,MoldawerLL.Cytokinesandwoundhealing:theroleof
cytokineandanticytokinetherapyintherepairresponse.The
JournalofBurncare&Rehabilitation,2004,
25
(2):149
-
160
[51]GutteridgeJM.IronpromotersoftheFentonreactionandlipid
peroxidationcanbereleasedfromhaemoglobinbyperoxides.
FEBSLetters,1986,
201
(2):291
-
295
[52]CemerskiS,CantagrelA,VanMeerwijkJP,
etal
.Reactiveoxygen
speciesdifferentiallyaffectTcellreceptor-signalingpathways.The
JournalofBiologicalChemistry,2002,
277
(22):19585
-
19593
[53]ZhuJ,ZouZZ,NiePP,
etal
.DownregulationofmicroRNA-
27b-3penhancestamoxifenresistanceinbreastcancerby
increasingNR5A2andCREB1expression.CellDeathDis,2016,
7
(11):e2454
[54]AoX,NiePP,WuBY,
etal
.DecreasedexpressionofmicroRNA-
17andmicroRNA-20bpromotesbreastcancerresistancetotaxol
therapybyupregulationofNCOA3.CellDeathDis,2016,
7
(11):
e2463
[55]ZouZZ,YuanZY,ZhangQX,
etal
.AurorakinaseAinhibition-
inducedautophagytriggersdrugresistanceinbreastcancercells.
Autophagy,2012,
8
(12):1798
-
1810
[56]RocksonSG.Lymphedema.AmericanJournalOfMedicine,2001,
110
(4):288
-
295
[57]SmootB,Chiavola-LarsonL,LeeJ,
etal
.Effectoflow-levellaser
therapyonpainandswellinginwomenwithbreastcancer-related
lymphedema:asystematicreviewandmeta-analysis.JCancer
Surviv,2015,
9
(2):287
-
304
[58]RidnerSH,Poage-HooperE,KanarC,
etal
.Apilotrandomized
trialevaluatinglow-levellasertherapyasanalternativetreatmentto
manuallymphaticdrainageforbreastcancer-relatedlymphedema.
OncolNursForum,2013,
40
(4):383
-
393
[59]HwangWT,ChungSH,LeeJS.Complexdecongestivephysical
therapyandlow-levellasertherapyforthetreatmentofpediatric
congenitallymphedema:acasereport.JPhysTherSci,2015,
27
(6):2021
-
2022
[60]KozanogluE,BasaranS,PaydasS,
etal
.Efficacyofpneumatic
compressionandlow-levellasertherapyinthetreatmentof
postmastectomylymphoedema:arandomizedcontrolledtrial.
ClinicalRehabilitation,2009,
23
(2):117
-
124
[61]OremusM,DayesI,WalkerK,
etal
.Systematicreview:
conservativetreatmentsforsecondarylymphedema.BMCCancer,
2012,
12
:6
[62]CaratiCJ,AndersonSN,GannonBJ,
etal
.Treatmentof
postmastectomylymphedemawithlow-levellasertherapy:adouble
blind,placebo-controlledtrial.Cancer,2003,
98
(6):1114
-
1122
[63]AgaibyAD,GhaliLR,WilsonR,
etal
.Lasermodulationof
angiogenicfactorproductionbyT-lymphocytes.LasersinSurgery
andMedicine,2000,
26
(4):357
-
363
[64]FengJ,ZhangYJ,XingD.Low-powerlaserirradiation(LPLI)
promotesVEGFexpressionandvascularendothelialcell
proliferationthroughtheactivationofERK/Sp1pathway.Cellular
Signalling,2012,
24
(6):1116
-
1125
[65]ParkIS,ChungPS,AhnJC.Adipose-derivedstemcellspheroid
treatedwithlow-levellightirradiationacceleratesspontaneous
angiogenesisinmousemodelofhindlimbischemia.Cytotherapy,
2017,
19
(9):1070
-
1078
[66]CauwelsRG,MartensLC.Lowlevellasertherapyinoral
mucositis:apilotstudy.EuropeanArchivesofPaediatricDentistry:
OfficialJournaloftheEuropeanAcademyofPaediatricDentistry,
2011,
12
(2):118
-
123
[67]GouveaDeLimaA,VillarRC,DeCastroG,Jr,
etal
.Oral
mucositispreventionbylow-levellasertherapyinhead-and-neck
cancerpatientsundergoingconcurrentchemoradiotherapy:aphase
IIIrandomizedstudy.InternationalJournalofRadiationOncology,
Biology,Physics,2012,
82
(1):270
-
275
[68]Arbabi-KalatiF,Arbabi-KalatiF,MoridiT.Evaluationoftheeffect
oflowlevellaseronpreventionofchemotherapy-induced
mucositis.ActaMedicaIranica,2013,
51
(3):157
-
162
[69]GautamAP,FernandesDJ,VidyasagarMS,
etal
.Lowlevellaser
therapyagainstradiationinducedoralmucositisinelderlyheadand
neckcancerpatients-arandomizedplacebocontrolledtrial.Journal
ofPhotochemistryandPhotobiologyB,Biology,2015,
144
:51
-
56
[70]OberoiS,Zamperlini-NettoG,BeyeneJ,
etal
.Effectof
prophylacticlowlevellasertherapyonoralmucositis:asystematic
reviewandmeta-analysis.PloSOne,2014,
9
(9):e107418
[71]AllanE,BarneyC,BaumS,
etal
.Low-levellasertherapyandlaser
1080· ·
好才,等:功率法对疫应影响
2017;44(12)
debridementformanagementoforalmucositisinpatientswith
headandneckcancerreceivingchemotherapyandradiation.IntJ
RadiatOncol,2016,
94
(4):883
[72]OngZY,GibsonRJ,BowenJM,
etal
.Pro-inflammatory
cytokinesplayakeyroleinthedevelopmentofradiotherapy-
inducedgastrointestinalmucositis.RadiationOncology,2010,
5
:22
[73]LoganR,StringerA,BowenJ,
etal
.Isthepathobiologyof
chemotherapy-inducedalimentarytractmucositisinfluencedbythe
typeofmucotoxicdrugadministered?CancerChemothPharm,
2009,
63
(2):239
-
251
[74]HamiltonS,YooJ,HammondA,
etal
.Microvascularchangesin
radiation-inducedoralmucositis.JournalofOtolaryngology-Head
&NeckSurgery
=
LeJournald'oto-rhino-laryngologieetde
chirurgiecervico-faciale,2008,
37
(5):730
-
737
[75]Al-DasooqiN,BowenJM,GibsonRJ,
etal
.Irinotecan-induced
alterationsinintestinalcellkineticsandextracellularmatrix
componentexpressionintheDarkAgoutirat.InternationalJournal
ofExperimentalPathology,2011,
92
(5):357
-
365
[76]SonisST.Thepathobiologyofmucositis.NatureReviewsCancer,
2004,
4
(4):277
-
284
[77]SilvaGBL,SaconoNT,Othon-LeiteAF,
etal
.Effectof
low-levellasertherapyoninflammatorymediatorreleaseduring
chemotherapy-inducedoralmucositis:arandomizedpreliminary
study.LasersinMedicalScience,2015,
30
(1):117
-
126
[78]Oton-LeiteAF,SilvaGBL,MoraisMO,
etal
.Effectoflow-level
lasertherapyonchemoradiotherapy-inducedoralmucositisand
salivaryinflammatorymediatorsinheadandneckcancerpatients.
LasersinSurgeryandMedicine,2015,
47
(4):296
-
305
[79]LopesNNF,PlaplerH,ChavantesMC,
etal
.Cyclooxygenase-2
andvascularendothelialgrowthfactorexpressionin
5-fluorouracil-inducedoralmucositisinhamsters:evaluationof
twolow-intensitylaserprotocols.SupportiveCareInCancer,2009,
17
(11):1409
-
1415
[80]LopesNNF,PlaplerH,LallaRV,
etal
.Effectsoflow-levellaser
therapyoncollagenexpressionandneutrophilinfiltratein
5-fluorouracil-inducedoralmucositisinhamsters. Lasersin
SurgeryandMedicine,2010,
42
(6):546
-
552
[81]CensabellaS,ClaesS,RobijnsJ,
etal
.Photobiomodulationforthe
managementofradiationdermatitis:theDERMIStrial,apilot
studyofMLS(R)lasertherapyinbreastcancerpatients.Supportive
CareInCancer,2016,
24
(9):3925
-
3933
[82]DelandMM,WeissRA,McdanielDH,
etal
.Treatmentof
radiation-induceddermatitiswithlight-emittingdiode(LED)
photomodulation.LasersinSurgeryandMedicine,2007,
39
(2):
164
-
168
[83]SchindlA,SchindlM,SchindlL,
etal
.Increaseddermal
angiogenesisafterlow-intensitylasertherapyforachronicradiation
ulcerdeterminedbyavideomeasuringsystem.JAmAcad
Dermatol,1999,
40
(3):481
-
484
[84]SchindlM,KerschanK,SchindlA,
etal
.Inductionofcomplete
woundhealinginrecalcitrantulcersbylow-intensitylaser
irradiationdependsonulcercauseandsize.PhotodermatolPhoto,
1999,
15
(1):18
-
21
[85]IhsanFRM.Low-levellasertherapyacceleratescollateral
circulationandenhancesmicrocirculation.Photomedicineand
LaserSurgery,2005,
23
(3):289
-
294
[86]ChawlaK,LambaAK,TandonS,
etal
.Effectoflow-levellaser
therapyonwoundhealingafterdepigmentationprocedure:A
clinicalstudy.JournalofIndianSocietyofPeriodontology,2016,
20
(2):184
-
188
[87]deLimaFM,AlbertiniR,DantasY,
etal
.Low-levellasertherapy
restorestheoxidativestressbalanceinacutelunginjuryinducedby
gutischemiaandreperfusion.PhotochemPhotobiol,2013,
89
(1):
179
-
188
[88]Ushio-FukaiM,AlexanderRW.Reactiveoxygenspeciesas
mediatorsofangiogenesissignaling-RoleofNAD(P)Hoxidase.
MolecularandCellularBiochemistry,2004,
264
(1
-
2):85
-
97
[89]IrwinDC,MccordJM,Nozik-GrayckE,
etal
.Apotentialrolefor
reactiveoxygenspeciesandtheHIF-1alpha-VEGFpathwayin
hypoxia-inducedpulmonaryvascularleak.FreeRadicalBioMed,
2009,
47
(1):55
-
61
[90]HeY,FanJ,LinH,
etal
.Theanti-malariaagentartesunateinhibits
expressionofvascularendothelialgrowthfactorandhypoxia-
induciblefactor-1alphainhumanrheumatoidarthritisfibroblast-
likesynoviocyte.RheumatologyInternational,2011,
31
(1):53
-
60
[91]MolitorisKH,KaziAA,KoosRD.Inhibitionofoxygen-induced
hypoxia-induciblefactor-1alphadegradationunmasksestradiol
inductionofvascularendothelialgrowthfactorexpressionin
ECC-1cancercells
invitro
.Endocrinology,2009,
150
(12):5405
-
5414
[92]GaoXJ,ChenTS,XingD,
etal
.SinglecellanalysisofPKC
activationduringproliferationandapoptosisinducedbylaser
irradiation.JournalofCellularPhysiology,2006,
206
(2):441
-
448
[93]HuangL,JiangXX,GongLL,
etal
.Photoactivationof
Akt1/GSK3isoform-specificsignalingaxispromotespancreatic-
cellregeneration.JournalofCellularBiochemistry,2015,
116
(8):
1741
-
1754
[94]BassoFG,SoaresDG,PansaniTN,
etal
.Proliferation,migration,
andexpressionoforal-mucosal-healing-relatedgenesbyoral
fibroblastsreceivinglow-levellasertherapyafterinflammatory
cytokineschallenge.LasersinSurgeryandMedicine,2016,
48
(10):
1006
-
1014
[95]BensadounRJ,NairRG.Low-levellasertherapyinthe
managementofmucositisanddermatitisinducedbycancertherapy.
PhotomedicineandLaserSurgery,2015,
33
(10):487
-
491
[96]LuCX,FanZJ,XingD.Photo-enhancementofmacrophage
phagocyticactivity
via
Rac1-mediatedsignalingpathway:
Implicationsforbacterialinfection.IntJBiochemCellB,2016,
78
:206
-
216
[97]KarunarathneWKA,GiriL,PatelAK,
etal
.Opticalcontrol
demonstratesswitch-likePIP3dynamicsunderlyingtheinitiationof
immunecellmigration.ProcNatlAcadSciUSA,2013,
110
(17):
E1575
-
E1583
1081· ·
化学物理进展 Prog.Biochem.Biophys. 2017;44(12)
功率照疗对机免疫应的影响
*
好才 1) 真真 2) 刘镭
1)**
(
1) 师范学生光子学究院光生科学究所暨激生命学教部重点验室广州
510631
2) 大学山医院人病毒究所,广州
510080)
摘要 近几年,采红至红外波长(6001100nm)的低功率光照(low-doselightLDL)疗法对织代谢系神经统、
系统等方节效引起广关注物能生物究的发展推了低功率
光照在疾领域革新指出细胞细胞、中粒细胞细胞免疫细胞应低率光照,
胞因子和护性白质分缓解些疾病的进程因此将从分子、细胞和组织水平低功率光照改善一些
疫学进行总结
键词 低功照,症反应,巴水肿咽黏膜炎性皮炎肿瘤抗菌
类号 R454 DOI:10.16476/j.pibb.2017.0275
[98]RudikDV,TikhomirovaEI.Activityofmurineperitoneal
macrophagesuponweakredandinfraredlaserirradiation
invitro
and
invivo
.Biophysics,2007,
52
(5):504
-
507
[99]CerdeiraCD,BrigagaoMRPL,DeCarliML,
etal
.Low-level
lasertherapystimulatestheoxidativeburstinhumanneutrophils
andincreasestheirfungicidalcapacity.JournalofBiophotonics,
2016,
9
(11
-
12):1180
-
1188
[100]EzzatiA,BayatM,KhoshvaghtiA.Low-levellasertherapywitha
pulsedinfraredlaseracceleratessecond-degreeburnhealinginrat:
aclinicalandmicrobiologicstudy.PhotomedicineandLaser
Surgery,2010,
28
(5):603
-
611
[101]SantosNRS,SobrinhoJBD,AlmeidaPF,
etal
.Influenceofthe
combinationofinfraredandredlaserlightonthehealingof
cutaneouswoundsinfectedbystaphylococcusaureus.
PhotomedicineandLaserSurgery,2011,
29
(3):177
-
182
[102]HawkinsD,AbrahamseH.Effectofmultipleexposuresof
low-levellasertherapyonthecellularresponsesofwoundedhuman
skinfibroblasts.PhotomedicineandLaserSurgery,2006,
24
(6):
705
-
714
[103]FrigoL,FaveroGM,LimaHJC,
etal
.Low-levellaserirradiation
(InGaAlP-660nm)increasesfibroblastcellproliferationand
reducescelldeathinadose-dependentmanner.Photomedicineand
LaserSurgery,2010,
28
(S1):S151
-
S156
[104]WuSN,XingD.Intracellularsignalingcascadesfollowinglight
irradiation.LaserPhotonicsRev,2014,
8
(1):115
-
130
[105]LuCX,ZhouFF,WuSN,
etal
.Phototherapy-inducedantitumor
immunity:long-termtumorsuppressioneffects
via
photoinactivationofrespiratorychainoxidase-triggeredsuperoxide
anionburst.AntioxidRedoxSign,2016,
24
(5):249
-
262
[106]WeiY,XingD.Phototherapy-treatedapoptotictumorcellsinduce
pro-inflammatorycytokinesproductioninmacrophage.Twelfth
InternationalConferenceonPhotonicsandImaginginBiologyand
Medicine,2014,
7160
(2):71601S-71601S-7
[107]ZhouF,XingD.HF-LPLI-treatedtumorcellsinduceNO
productioninmacrophage.ProcSPIE,2013,
8582
(16):7629
-
7638
[108]SonisST,HashemiS,EpsteinJB,
etal
.Couldthebiological
robustnessoflowlevellasertherapy(Photobiomodulation)impact
itsuseinthemanagementofmucositisinheadandneckcancer
patients.OralOncology,2016,
54
:7
-
14
*
自然学基
(61361160414,31470072,61405061)
和广自然学基
(2014A030313419)
资助项目
.
**
通讯联系人
.
Tel:020-85211436, E-mail:liulei@scnu.edu.cn
稿日期:
2017-07-13
期:
2017-10-19
1082· ·
Article
Full-text available
Melanoma with rapid progression towards metastasis has become the deadliest form of skin cancer. However, the mechanism of melanoma growth and metastasis is still unclear. Here, we found that miRNA-138 was lowly expressed and hypoxia-inducible factor 1α (HIF1α) was highly expressed in patients' melanoma tissue compared with the paracancerous tissues, and they had a significant negative correlation (r=-0.877, P<0.001). Patients with miRNA-138low/HIF1αhigh signatures were predominant in late stage III/IV of melanoma. Further, bioinformatic analysis demonstrated that miRNA-138 directly targeted HIF1α. We found that the introduction of pre-miRNA-138 sequences to A375 cells reduced HIF1α mRNA expression and suppressed cell proliferation, migration and invasion. Overexpression of miRNA-138 or inhibition of HIF1α significantly suppressed the growth and metastasis of melanoma in vivo Our study demonstrates the role and clinical relevance of miRNA-138 and HIF1α in melanoma cell growth and metastasis, providing a novel therapeutic target for suppression of melanoma growth and metastasis.
Article
Vascular endothelial growth factor (VEGF) signaling promotes angiogenesis by stimulating the migration and proliferation of endothelial cells. The aim of this study was to investigate the expression of Survivin and VEGF receptor 1/2/3 (VEGFR 1/2/3) in esophageal carcinoma tissues (ECTs), and to explore the therapy effect of the suppression of VEGFR2 signaling. Here, we found that VEGFR2 and Survivin had high expressions and a significant correlation (r = 0.874, P < 0.002) in ECTs. Further, we found that VEGFR2 signaling could activate the AKT1/MDM2/Survivin pathway. The inhibition of VEGFR2 signaling with the XL184 treatment downregulated the phosphorylation of AKT1 and MDM2, and then, increased the activation of Caspase 3/7, resulting in the reduction of cell viability and the apoptosis of HUVECs. Additionally, in the esophageal tumor model, the tumor growth was significantly suppressed by blocking Survivin and the suppression of tumor growth was more effective in the combined treatment by blocking Survivin and Bcl-xl/Bcl-2. Our data thus revealed that Survivin in the signal downstream of VEGFR2 played an important role in esophageal cancer cell survival and might be a potential candidate target for the combined therapy for esophageal cancer.
Article
Full-text available
To evaluate the efficacy of low-level laser therapy (LLLT) on collateral circulation and microcirculation if a blood vessel is occluded. Investigators have attempted prostaglandin and ultrasound therapy to promote improvements in the vascular bed of deprived tissue after an injury, which may lead to occlusion of the blood vessels. Thirty-four adult rabbits were used in this study, two of them considered 0-h reading group, while the rest were divided into two equal groups, with 16 rabbits each: control and those treated with LLLT. Each rabbit underwent two surgical operations; the medial aspect of each thigh was slit, the skin incised and the femoral artery exposed and ligated. The site of the operation in the treated group was irradiated directly following the operation and for 3 d after, one session daily for 10 min/session. The laser system used was a gallium-aluminum-arsenide (Ga-Al-As) diode laser with a wavelength of 904 nm and power of 10 mW. Blood samples collected from the femoral artery above the site of the ligation were sent for examination with high-performance liquid chromatography (HPLC) to determine the levels of adenosine, growth hormone (GH) and fibroblast growth factor (FGF). Tissue specimens collected from the site of the operation, consisting of the artery and its surrounding muscle fibers, were sent for histopathological examination to determine the fiber/capillary (F/C) ratio and capillary diameter. Blood samples and tissue specimens were collected at 4, 8, 12, 16, 20, 24, 48 and 72 h postoperatively from the animals of both groups, control and treated. Rapid increases in the level of adenosine, GH, and FGF occurred. The F/C ratio and capillary diameter peaked at 12-16 h; their levels declined gradually, reaching normal values 72 h after irradiation in the treated group. Numerous collateral blood vessels proliferated the area, with marked increases in the diameters of the original blood vessels. The results indicated that LLLT accelerated collateral circulation and enhanced microcirculation and seemed to be unique in the normalization of the functional features of the injured area, which could lead to occlusion of the regional blood vessels.
Article
Full-text available
Macrophages and resident microglia play an import role in the secondary neuroinflammation response following spinal cord injury. Reprogramming of macrophage/microglia polarization is an import strategy for spinal cord injury restoration. Low-level laser therapy (LLLT) is a noninvasive treatment that has been widely used in neurotrauma and neurodegenerative diseases. However, the influence of low-level laser on polarization of macrophage/microglia following spinal cord injury remains unknown. The present study applied low-level laser therapy on a crush spinal cord injury rat model. Using immunofluorescence, flow cytometry, RT-qPCR, and western blot assays, we found that low-level laser therapy altered the polarization state to a M2 tendency. A greater number of neurons survived in the pare injury site, which was accompanied by higher BBB scores in the LLLT group. Furthermore, low-level laser therapy elevated expression of interleukin 4 (IL-4) and interleukin 13 (IL-13). Results from this study show that low-level laser therapy has the potential for reducing inflammation, regulating macrophage/microglia polarization, and promoting neuronal survival. These beneficial effects demonstrate that low-level laser therapy may be an effective candidate for clinical treatment of spinal cord injury.
Article
Full-text available
Chemoresistance is a major obstacle to effective breast cancer chemotherapy. However, the underlying molecular mechanisms remain unclear. In this study, nuclear receptor coactivator 3 (NCOA3) was found to be significantly increased in taxol-resistant breast cancer tissues and cells. Moreover, overexpression of NCOA3 enhanced breast cancer cell resistance to taxol, whereas depletion of NCOA3 decreased taxol resistance. Subsequently, we investigated whether NCOA3 expression was regulated by miRNAs in breast cancer. By bioinformatics prediction in combination with the data of previous report, miR-17 and miR-20b were selected as the potential miRNAs targeting NCOA3. By real-time PCR analysis, we found that miR-17 and miR-20b were significantly reduced in taxol-resistant breast cancer tissues and cells. In addition, we provided some experimental evidences that miR-17 and miR-20b attenuated breast cancer resistance to taxol in vitro and in vivo models. Furthermore, by luciferase reporter assays, we further validated that both miR-17 and miR-20b directly binded the 3′-untranslated region of NCOA3 mRNA and inhibited its expression in breast cancer cells. Finally, both miR-17 and miR-20b levels were found to be significantly negatively correlated with NCOA3 mRNA levels in breast cancer tissues. Together, our results indicated that loss of miR-17 and miR-20b enhanced breast cancer resistance to taxol by upregulating NCOA3 levels. Our study suggested miR-17, miR-20b and NCOA3 may serve as some predictive biomarkers and potential therapeutic targets in taxol-resistant breast cancer treatment.
Article
Full-text available
Estrogen-dependent breast cancer is often treated with the aromatase inhibitors or estrogen receptor (ER) antagonists. Tamoxifen as a major ER antagonist is usually used to treat those patients with ERα-positive breast cancer. However, a majority of patients with ERα positive fail to respond to tamoxifen due to the presence of intrinsic or acquired resistance to the drug. Altered expression and functions of microRNAs (miRNAs) have been reportedly associated with tamoxifen resistance. In this study, we investigated the role of miR-27b-3p in resistance of breast cancer to tamoxifen. MiR-27b-3p levels were remarkably reduced in the tamoxifen-resistant breast cancer cells compared with their parental cells. In addition, miR-27b-3p was also significantly downregulated in breast tumor tissues relative to adjacent non-tumor tissues. Moreover, the expression levels of miR-27b-3p were lower in the breast cancer tissues from tamoxifen-resistant patients compared with that from untreated-tamoxifen patients. Notably, tamoxifen repressed miR-27b-3p expression, whereas estrogen induced miR-27b-3p expression in breast cancer cells. Besides, we provided experimental evidences that miR-27b-3p enhances the sensitivity of breast cancer cells to tamoxifen in vitro and in vivo models. More importantly, we validated that miR-27b-3p directly targeted and inhibited the expression of nuclear receptor subfamily 5 group A member 2 (NR5A2) and cAMP-response element binding protein 1 (CREB1) and therefore augmented tamoxifen-induced cytotoxicity in breast cancer. Lastly, miR-27b-3p levels were found to be significantly negatively correlated with both NR5A2 and CREB1 levels in breast cancer tissues. Our findings provided further evidence that miR-27b-3p might be considered as a novel and potential target for the diagnosis and treatment of tamoxifen-resistant breast cancer.
Article
Full-text available
Background and objectives: Increased expression of inflammatory cytokines in the oral cavity has been related to the etiopathogenesis of oral mucositis and to delayed oral mucosal repair. Low-level laser therapy (LLLT) stimulates proliferation and migration of gingival fibroblasts, but the effects of specific inflammatory cytokines on oral mucosal cells and the modulation of these effects by LLLT have not been fully investigated. Therefore, this study investigated the effects of LLLT on oral fibroblasts after being challenged by oral-mucositis-related inflammatory cytokines. Methods: Human gingival fibroblasts were seeded in plain culture medium (DMEM) containing 10% fetal bovine serum (FBS) for 24 hours. Then, cells were kept in contact with inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-8) in serum-free DMEM for 24 hours. After this period, cells were subjected to LLLT with a diode laser device (LaserTABLE, InGaAsP, 780 nm, 25 mW) delivering energy doses from 0.5 to 3 J/cm2 . Irradiation was repeated for 3 consecutive days. Twenty-four hours after the last irradiation, cell migration (wound-healing and transwell migration assays), cell proliferation (BrdU), gene expression of COL-I and growth factors (real-time PCR), and synthesis of COL-I (Sirius Red assay) and VEGF (ELISA) were assessed. Data were subjected to two-way ANOVA and Tukey's tests or Kruskall-Walis and Mann-Whitney tests (P < 0.05). Results: The inflammatory cytokines decreased the migration capacity of gingival fibroblasts. However, a statistically significant difference was observed only for IL-6, detected by transwell assay, where 30% less cells migrated through the pores (P < 0.05) and IL-8, with an increased wound area (116%; P < 0.05), detected by the wound healing method. Cell proliferation was not affected by contact with cytokines, while growth factors and COL-I expression (approximately 80%; P < 0.05), as well as VEGF synthesis (approximately 20%; P < 0.05), were decreased after contact to all tested cytokines. The opposite was seen for total collagen synthesis. LLLT promoted an acceleration of fibroblast migration (30%; P < 0.05) and proliferation (112%; P < 0.05) when delivering 0.5 J/cm2 to the cells previously in contact with the inflammatory cytokines. Gene expression of VEGF (approximately 30%; P < 0.05), and EGF (17%; P < 0.05), was stimulated by LLLT after contact with TNF-α and IL-6. Conclusion: LLLT can counteract the negative effects of high concentrations of inflammatory cytokines, especially IL-6 and IL-8 on gingival fibroblast functions directly related to the wound-healing process. Lasers Surg. Med. 48:1006-1014, 2016. © 2016 Wiley Periodicals, Inc.
Article
Full-text available
Phagocytosis and the subsequent destruction of invading pathogens by macrophages are indispensable steps in host immune responses to microbial infections. Low-power laser irradiation (LPLI) has been found to exert photobiological effects on immune responses, but the signaling mechanisms underlying this photobiomodulation of phagocytosis remain largely unknown. Here, we demonstrated for the first time that LPLI enhanced the phagocytic activity of macrophages by stimulating the activation of Rac1. The overexpression of constitutively activated Rac1 clearly enhanced LPLI-induced phagocytosis, whereas the overexpression of dominant negative Rac1 exerted the opposite effect. The phosphorylation of cofilin was involved in the effects of LPLI on phagocytosis, which was regulated by the membrane translocation and activation of Rac1. Furthermore, the photoactivation of Rac1 was dependent on the Src/PI3K/Vav1 pathway. The inhibition of the Src/PI3K pathway significantly suppressed LPLI-induced actin polymerization and phagocytosis enhancement. Additionally, LPLI-treated mice exhibited increased survival and a decreased organ bacterial load when challenged with Listeria monocytogenes, indicating that LPLI enhanced macrophage phagocytosis in vivo. These findings highlight the important roles of the Src/PI3K/Vav1/Rac1/cofilin pathway in regulating macrophage phagocytosis and provide a potential strategy for treating phagocytic deficiency via LPLI.
Article
Full-text available
Purpose The aim of this study was to assess the effectiveness and acceptability of photobiomodulation using MLS® laser therapy (LT) in the management of acute radiation dermatitis (RD). Methods We compared two successive groups of breast cancer patients undergoing identical radiotherapy regimens post-lumpectomy. Both groups received our standard skin care but the second group received six additional LT sessions (beam area 19.635 cm2, 0.168 W/cm2, 4 J/cm2), starting at fraction 20 of radiotherapy (control and LT group, N = 41 and 38, respectively). The clinical outcomes were the severity of RD (using the Radiation Therapy Oncology Group [RTOG] criteria and the Radiotherapy-Induced Skin Reaction Assessment Scale [RISRAS]) and dermatology-specific quality of life (Skindex-16) before the start of LT and at the end of radiotherapy. Secondary outcomes were patients’ ratings of skin care or LT (pleasantness, soothing effect, and global satisfaction). Results Skin toxicity was equivalent between the groups before the start of LT but significantly differed at the end of radiotherapy, with an aggravation in the control but not in the LT group (e.g., 29 versus 3 % of RTOG grade 2 RD, respectively, P < 0.005). We found no significant group differences with respect to quality of life. However, the RISRAS subjective score decreased in the LT group only, implying a decreased impact of RD on patients’ quality of life. Finally, patients’ ratings were significantly higher for LT than for standard care. Conclusions These findings suggest that LT might be effective to manage acute RD and warrant further research. Trial registration Clinical trial number: NCT01932073. https:// clinicaltrials. gov/ ct2/ show/ NCT01932073.
Article
Background aims: We investigated whether low-level light irradiation (LLLI) before adipose-derived stromal cells (ASCs) spheroid transplantation improved hind-limb functional recovery by stimulation of angiogenesis. Methods: The spheroid, composed of ASCs, was irradiated with low-level light and expressed angiogenic factors, including vascular endothelial growth factor and basic fibroblast growth factor. From immunochemical staining analysis, the spheroid of ASCs included CD31(+), KDR(+) and CD34(+), whereas monolayer-cultured ASCs were negative for these markers. To evaluate the therapeutic effect of the ASC spheroid treated with LLLI in vivo, phosphate-buffered saline, monolayer ASCs, LLLI-monolayer ASCs, spheroid ASCs and LLLI-spheroid ASCs were transplanted into a hind-limb ischemia model. Results: The LLLI-spheroid ASCs transplanted into the hind-limb ischemia differentiated into endothelial cells and remained differentiated. Transplantation of LLLI-spheroid ASCs into the hind-limb ischemia significantly elevated the density of vascular formations through angiogenic factors released by the ASCs and enhanced tissue regeneration at the lesion site. Consistent with these results, the transplantation of LLLI-spheroid ASCs significantly improved functional recovery compared with ASC or spheroid ASC transplantation and PBS treatment. Conclusions: These findings suggest that transplantation of ASC spheroid treated with LLLI may be an effective stem cell therapy for the treatment of hind-limb ischemia and peripheral vascular disease.
Article
Low‐level laser therapy (LLLT) is known to enhance mitochondrial electron transfer and ATP production; thus, this study asked whether LLLT could stimulate the oxidative burst in human neutrophils (PMN) and improve their ability to kill microorganisms. Blood from healthy human subjects was collected and PMN were isolated from the samples. PMN were treated in vitro with 660 nm or 780 nm CW laser light at 40 mW power and increasing energies up to 19.2 J and were subsequently incubated with Candida albicans cells. Generation of hydroxyl radicals, hypochlorite anions and superoxide anions by PMN were checked using fluorescent probes and chemiluminescence assays; a microbicidal activity assay against C. albicans was also performed. LLLT excited PMN to a higher functional profile, which was translated as superior production of reactive oxygen species (ROS) and increased fungicidal capacity. The most efficacious energy was 19.2 J and, interestingly, the 660 nm light was even more efficacious than 780 nm at increasing the respiratory burst of PMN and the fungicidal capacity. Human neutrophils (PMN) were stimulated in vitro with 660 nm or 780 nm CW laser light at 40 mW of power and a total energy of 19.2 J. Low‐level laser therapy (LLLT) excited PMN to a higher functional profile, which was translated as a superior production of reactive oxygen species (ROS) such as hydroxyl radicals (HO • ) and hypochlorite anions (ClO ⁻ ) (Figure) and increased fungicidal capacity against Candida albicans cells. magnified image Human neutrophils (PMN) were stimulated in vitro with 660 nm or 780 nm CW laser light at 40 mW of power and a total energy of 19.2 J. Low‐level laser therapy (LLLT) excited PMN to a higher functional profile, which was translated as a superior production of reactive oxygen species (ROS) such as hydroxyl radicals (HO • ) and hypochlorite anions (ClO ⁻ ) (Figure) and increased fungicidal capacity against Candida albicans cells.